Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Oncotarget ; 8(15): 25115-25130, 2017 Apr 11.
Article in English | MEDLINE | ID: mdl-28445989

ABSTRACT

TMPRSS2:ERG (T/E) gene fusions are present in approximately 50% of all prostate cancer (PCa) cases. The expression of fusion mRNAs from distinct T/E variants is associated with clinicopathological parameters, while the underlying molecular processes remain unclear. We characterized the molecular mechanisms and functional implications caused by doxycycline (Dox)-inducible overexpression of the frequent T/E III and VI fusion variants in LNCaP cells. Induction of T/E expression resulted in increased cellular migratory and invasive potential, and reduced proliferation and accumulation in G1 phase. T/E overexpressing cells showed epithelial-to-mesenchymal transition (EMT), as demonstrated by upregulation of TGF-ß and WNT pathway genes, mesenchymal markers, and increased phosphorylation of the p38 MAPK. Augmented secretion of TGF-ß1 and -ß2, and T/E-mediated regulation of ALK1, a member of the TGF-ß receptor family, was detected. ALK1 inhibition in T/E overexpressing cells blocked p38 phosphorylation and reduced the expression of the TGF-ß target genes VIM, MMP1, CDH2, and SNAI2. We found a T/E variant VI-specific induction of miR-503 associated with reduced expression of SMAD7 and CDH1. Overexpression of miR-503 led to increased levels of VIM and MMP1. Our findings indicate that TGF-ß signaling is a major determinant of EMT in T/E overexpressing LNCaP cells. We provide evidence that T/E VI-specific transcriptional modulation by miR-503 accounts for differences in the activation of EMT pathway genes, promoting the aggressive phenotype of tumors expressing T/E variant VI. We suggest that ALK1-mediated TGF-ß signaling is a novel oncogenic mechanism in T/E positive PCa.


Subject(s)
Epithelial-Mesenchymal Transition/genetics , Oncogene Proteins, Fusion/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Serine Endopeptidases/genetics , Signal Transduction , Transforming Growth Factor beta/metabolism , Activin Receptors, Type II/antagonists & inhibitors , Activin Receptors, Type II/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Genetic Variation , Humans , Male , Models, Biological , Prostatic Neoplasms/pathology , Signal Transduction/drug effects , Smad7 Protein/antagonists & inhibitors , Smad7 Protein/metabolism , Transcription, Genetic , Transcriptional Regulator ERG/genetics , beta Catenin/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
2.
Immunogenetics ; 69(6): 401-407, 2017 06.
Article in English | MEDLINE | ID: mdl-28364129

ABSTRACT

The Scavenger Receptor Cysteine-Rich (SRCR) proteins are an archaic group of proteins characterized by the presence of multiple SRCR domains. They are membrane-bound or secreted proteins, which are generally related to host defense systems in animals. Deleted in Malignant Brain Tumors 1 (DMBT1) is a SRCR protein which is secreted in mucosal fluids and involved in host defense by pathogen binding by its SRCR domains. Genetic polymorphism within DMBT1 leads to DMBT1-alleles giving rise to polypeptides with interindividually different numbers of SRCR domains, ranging from 8 SRCR domains (encoded by 6 kb DMBT1 variant) to 13 SRCR domains (encoded by the 8 kb DMBT1 variant). In the present study, we have investigated whether reduction from 13 to 8 amino-terminal SRCR domains leads to reduction of bacterial binding. The 6 kb variant bound ~20-45% less bacteria compared to the 8 kb variant. These results support the hypothesis that genetic variation in DMBT1 may influence microbial defense.


Subject(s)
Germ-Line Mutation , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Receptors, Scavenger/genetics , Receptors, Scavenger/metabolism , Sequence Deletion , Bacterial Adhesion/genetics , Calcium-Binding Proteins , DNA-Binding Proteins , Humans , Polymorphism, Genetic , Protein Binding , Protein Interaction Domains and Motifs , Receptors, Cell Surface/chemistry , Receptors, Scavenger/chemistry , Tumor Suppressor Proteins
3.
Oncotarget ; 8(14): 23760-23774, 2017 Apr 04.
Article in English | MEDLINE | ID: mdl-28423600

ABSTRACT

Next-generation sequencing has dramatically increased genome-wide profiling options and conceptually initiates the possibility for personalized cancer therapy. State-of-the-art sequencing studies yield large candidate gene sets comprising dozens or hundreds of mutated genes. However, few technologies are available for the systematic downstream evaluation of these results to identify novel starting points of future cancer therapies.We improved and extended a site-specific recombination-based system for systematic analysis of the individual functions of a large number of candidate genes. This was facilitated by a novel system for the construction of isogenic constitutive and inducible gain- and loss-of-function cell lines. Additionally, we demonstrate the construction of isogenic cell lines with combinations of the traits for advanced functional in vitro analyses. In a proof-of-concept experiment, a library of 108 isogenic melanoma cell lines was constructed and 8 genes were identified that significantly reduced viability in a discovery screen and in an independent validation screen. Here, we demonstrate the broad applicability of this recombination-based method and we proved its potential to identify new drug targets via the identification of the tumor suppressor DUSP6 as potential synthetic lethal target in melanoma cell lines with BRAF V600E mutations and high DUSP6 expression.


Subject(s)
Dual Specificity Phosphatase 6/genetics , Melanoma/genetics , Cell Line, Tumor , Dual Specificity Phosphatase 6/biosynthesis , Dual Specificity Phosphatase 6/metabolism , Gene Knockdown Techniques , Humans , Mass Screening , Melanoma/metabolism , Melanoma/pathology , Mutation , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Recombination, Genetic , Transfection
4.
Physiol Rev ; 96(2): 409-47, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26842265

ABSTRACT

Cartilage and bone are severely affected by glucocorticoids (GCs), steroid hormones that are frequently used to treat inflammatory diseases. Major complications associated with long-term steroid therapy include impairment of cartilaginous bone growth and GC-induced osteoporosis. Particularly in arthritis, GC application can increase joint and bone damage. Contrarily, endogenous GC release supports cartilage and bone integrity. In the last decade, substantial progress in the understanding of the molecular mechanisms of GC action has been gained through genome-wide binding studies of the GC receptor. These genomic approaches have revolutionized our understanding of gene regulation by ligand-induced transcription factors in general. Furthermore, specific inactivation of GC signaling and the GC receptor in bone and cartilage cells of rodent models has enabled the cell-specific effects of GCs in normal tissue homeostasis, inflammatory bone diseases, and GC-induced osteoporosis to be dissected. In this review, we summarize the current view of GC action in cartilage and bone. We further discuss future research directions in the context of new concepts for optimized steroid therapies with less detrimental effects on bone.


Subject(s)
Bone and Bones/drug effects , Cartilage/drug effects , Gene Expression Regulation , Glucocorticoids/adverse effects , Receptors, Glucocorticoid/metabolism , Animals , Arthritis, Rheumatoid/drug therapy , Bone Remodeling/drug effects , Disease Models, Animal , Growth Plate/drug effects , Humans , Insulin Resistance , Mesenchymal Stem Cells/drug effects , Osteoarthritis/drug therapy , Receptor Cross-Talk
5.
J Biomol Screen ; 18(6): 736-43, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23479354

ABSTRACT

In the past, the majority of antitumor compound-screening approaches had been performed in two-dimensional (2D) cell cultures. Although easy to standardize, this method provides results of limited significance because cells are surrounded by an artificial microenvironment, are not exposed to hypoxia gradients, and lack cell-cell contacts. These nonphysiological conditions directly affect relevant parameters such as the resistance to anticancer drugs. Multicellular tumor spheroids more closely resemble the in vivo situation in avascularized tumors. To monitor cellular reactions within this three-dimensional model system, we stably transfected a spheroid-forming glioblastoma cell line with Grx1-roGFP2, a green fluorescent protein (GFP)-based glutathione-specific redox sensor that detects alterations in the glutathione redox potential. Functionality and temporal dynamics of the sensor were verified with redox-active substances in 2D cell culture. Based on structured illumination microscopy using nonphototoxic light doses, ratio imaging was then applied to monitor the response of the glutathione system to exogenous hydrogen peroxide in optical sections of a tumor spheroid. Our approach provides a proof of concept for biosensor-based imaging in 3D cell cultures.


Subject(s)
Biosensing Techniques/methods , Cell Culture Techniques/methods , Drug Evaluation, Preclinical/methods , Imaging, Three-Dimensional/methods , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Glioblastoma/drug therapy , Glioblastoma/metabolism , Glutathione/metabolism , Green Fluorescent Proteins/metabolism , Humans , Hydrogen Peroxide/metabolism , Microscopy/methods , Oxidation-Reduction , Spheroids, Cellular/drug effects
6.
Neoplasia ; 13(7): 579-89, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21750652

ABSTRACT

The serine-protease hepsin is one of the most prominently overexpressed genes in human prostate carcinoma. Forced expression of the enzyme in mice prostates is associated with matrix degradation, invasive growth, and prostate cancer progression. Conversely, hepsin overexpression in metastatic prostate cancer cell lines was reported to induce cell cycle arrest and reduction of invasive growth in vitro. We used a system for doxycycline (dox)-inducible target gene expression in metastasis-derived PC3 cells to analyze the effects of hepsin in a quantitative manner. Loss of viability and adhesion correlated with hepsin expression levels during anchorage-dependent but not anchorage-independent growth. Full expression of hepsin led to cell death and detachment and was specifically associated with reduced phosphorylation of AKT at Ser(473), which was restored by growth on matrix derived from RWPE1 normal prostatic epithelial cells. In the chorioallantoic membrane xenograft model, hepsin overexpression in PC3 cells reduced the viability of tumors but did not suppress invasive growth. The data presented here provide evidence that elevated levels of hepsin interfere with cell adhesion and viability in the background of prostate cancer as well as other tissue types, the details of which depend on the microenvironment provided. Our findings suggest that overexpression of the enzyme in prostate carcinogenesis must be spatially and temporally restricted for the efficient development of tumors and metastases.


Subject(s)
Adenocarcinoma/genetics , Extracellular Matrix/physiology , Prostatic Neoplasms/genetics , Proto-Oncogene Proteins c-akt/genetics , Serine Endopeptidases/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Cell Line, Tumor , Chick Embryo , Extracellular Matrix/metabolism , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Neoplasm Invasiveness , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , Serine Endopeptidases/metabolism , Transfection , Transplantation, Heterologous , Up-Regulation/genetics
7.
Chem Commun (Camb) ; 47(1): 188-90, 2011 Jan 07.
Article in English | MEDLINE | ID: mdl-20830348
8.
Eur J Immunol ; 39(3): 833-42, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19189310

ABSTRACT

Deleted in malignant brain tumors 1 (DMBT1) is a secreted glycoprotein displaying a broad bacterial-binding spectrum. Recent functional and genetic studies linked DMBT1 to the suppression of LPS-induced TLR4-mediated NF-kappaB activation and to the pathogenesis of Crohn's disease. Here, we aimed at unraveling the molecular basis of its function in mucosal protection and of its broad pathogen-binding specificity. We report that DMBT1 directly interacts with dextran sulfate sodium (DSS) and carrageenan, a structurally similar sulfated polysaccharide, which is used as a texturizer and thickener in human dietary products. However, binding of DMBT1 does not reduce the cytotoxic effects of these agents to intestinal epithelial cells in vitro. DSS and carrageenan compete for DMBT1-mediated bacterial aggregation via interaction with its bacterial-recognition motif. Competition and ELISA studies identify poly-sulfated and poly-phosphorylated structures as ligands for this recognition motif, such as heparansulfate, LPS, and lipoteichoic acid. Dose-response studies in Dmbt1(-/-) and Dmbt1(+/+) mice utilizing the DSS-induced colitis model demonstrate a differential response only to low but not to high DSS doses. We propose that DMBT1 functions as pattern-recognition molecule for poly-sulfated and poly-phosphorylated ligands providing a molecular basis for its broad bacterial-binding specificity and its inhibitory effects on LPS-induced TLR4-mediated NF-kappaB activation.


Subject(s)
Carrageenan/immunology , Dextran Sulfate/immunology , Receptors, Cell Surface/immunology , Bacteria/immunology , Bacteria/metabolism , Calcium-Binding Proteins , Carrageenan/pharmacology , Carrageenan/toxicity , Cell Line , DNA-Binding Proteins , Dextran Sulfate/pharmacology , Dextran Sulfate/toxicity , Epithelial Cells/immunology , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Humans , Intestinal Mucosa/metabolism , Intestines/immunology , Intestines/microbiology , Ligands , Phosphates/immunology , Phosphates/metabolism , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Tumor Suppressor Proteins
9.
Gastroenterology ; 133(5): 1499-509, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17983803

ABSTRACT

BACKGROUND & AIMS: Impaired mucosal defense plays an important role in the pathogenesis of Crohn's disease (CD), one of the main subtypes of inflammatory bowel disease (IBD). Deleted in malignant brain tumors 1 (DMBT1) is a secreted scavenger receptor cysteine-rich protein with predominant expression in the intestine and has been proposed to exert possible functions in regenerative processes and pathogen defense. Here, we aimed at analyzing the role of DMBT1 in IBD. METHODS: We studied DMBT1 expression in IBD and normal tissues by quantitative reverse transcription-polymerase chain reaction, immunohistochemistry, and mRNA in situ hybridization. Genetic polymorphisms within DMBT1 were analyzed in an Italian IBD case-control sample. Dmbt1(-/-) mice were generated, characterized, and analyzed for their susceptibility to dextran sulfate sodium-induced colitis. RESULTS: DMBT1 levels correlate with disease activity in inflamed IBD tissues. A highly significant fraction of the patients with IBD displayed up-regulation of DMBT1 specifically in the intestinal epithelial surface cells and Paneth cells. A deletion allele of DMBT1 with a reduced number of scavenger receptor cysteine-rich domain coding exons is associated with an increased risk of CD (P = .00056; odds ratio, 1.75) but not for ulcerative colitis. Dmbt1(-/-) mice display enhanced susceptibility to dextran sulfate sodium-induced colitis and elevated Tnf, Il6, and Nod2 expression levels during inflammation. CONCLUSIONS: DMBT1 may play a role in intestinal mucosal protection and prevention of inflammation. Impaired DMBT1 function may contribute to the pathogenesis of CD.


Subject(s)
Crohn Disease/genetics , Crohn Disease/physiopathology , Gene Deletion , Intestinal Mucosa/physiopathology , Receptors, Cell Surface/genetics , Receptors, Cell Surface/physiology , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Calcium-Binding Proteins , Case-Control Studies , Child , Crohn Disease/chemically induced , DNA-Binding Proteins , Dextran Sulfate , Disease Susceptibility , Exons/genetics , Female , Humans , Interleukin-6/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Male , Mice , Mice, Transgenic , Middle Aged , Mucins/genetics , Mucins/physiology , Nod2 Signaling Adaptor Protein/metabolism , RNA, Messenger/metabolism , Risk Factors , Tumor Necrosis Factor-alpha/metabolism , Tumor Suppressor Proteins , Up-Regulation/genetics
10.
Electrophoresis ; 26(9): 1687-91, 2005 May.
Article in English | MEDLINE | ID: mdl-15812845

ABSTRACT

Combining multiplex reverse transcription-polymerase chain reaction (mRT-PCR) with microfluidic amplicon analysis, we developed an assay for the rapid and reliable semiquantitative expression screening of 11 candidate genes for drug resistance in human malignant melanoma. The functionality of this approach was demonstrated by low interexperimental variations of amplicon quantities after endpoint analysis. When applied to RNA samples derived from drug-sensitive and -resistant melanoma cell lines, mRT-PCR delivered results qualitatively concordant with data obtained from Northern blot and array analyses. The screening of additional melanoma cell lines resulted in distinct expression patterns for ten candidate genes. Our approach reveals a rapid and easy-to-handle alternative for candidate gene set evaluation from limited amounts of RNA.


Subject(s)
Drug Resistance, Neoplasm/genetics , Electrophoresis, Microchip , Genetic Testing/methods , Melanoma/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods , Cell Line, Tumor , Humans , Melanoma/diagnosis
11.
J Biol Chem ; 279(46): 47699-703, 2004 Nov 12.
Article in English | MEDLINE | ID: mdl-15355985

ABSTRACT

The scavenger receptor cysteine-rich (SRCR) proteins form an archaic group of metazoan proteins characterized by the presence of SRCR domains. These proteins are classified in group A and B based on the number of conserved cysteine residues in their SRCR domains, i.e. six for group A and eight for group B. The protein DMBT1 (deleted in malignant brain tumors 1), which is identical to salivary agglutinin and lung gp-340, belongs to the group B SRCR proteins and is considered to be involved in tumor suppression and host defense by pathogen binding. In a previous study we used nonoverlapping synthetic peptides covering the SRCR consensus sequence to identify a 16-amino acid bacteria-binding protein loop (peptide SRCRP2; QGRVEVLYRGSWGTVC) within the SRCR domains. In this study, using overlapping peptides, we pinpointed the minimal bacteria-binding site on SRCRP2, and thus DMBT1, to an 11-amino acid motif (DMBT1 pathogen-binding site 1 or DMBT1pbs1; GRVEVLYRGSW). An alanine substitution scan revealed that VEVL and Trp are critical residues in this motif. Bacteria binding by DMBT1pbs1 was different from the bacteria binding by the macrophage receptor MARCO in which an RXR motif was critical. In addition, the homologous consensus sequences of a number of SRCR proteins were synthesized and tested for bacteria binding. Only consensus sequences of DMBT1 orthologues bound bacteria by this motif.


Subject(s)
Agglutinins/genetics , Agglutinins/metabolism , Amino Acid Sequence , Bacteria/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Agglutinins/chemistry , Animals , Bacteria/pathogenicity , Binding Sites , Calcium-Binding Proteins , Consensus Sequence , DNA-Binding Proteins , Molecular Sequence Data , Peptides/genetics , Peptides/metabolism , Protein Binding , Protein Structure, Tertiary , Receptors, Cell Surface/chemistry , Sequence Alignment , Tumor Suppressor Proteins
12.
Genes Chromosomes Cancer ; 39(3): 185-94, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14732920

ABSTRACT

Deleted in malignant brain tumors 1 (DMBT1) has been proposed as a candidate tumor suppressor for brain and epithelial cancer. Initial studies suggested loss of expression rather than mutation as the predominant mode of DMBT1 inactivation. However, in situ studies in lung cancer demonstrated highly sophisticated changes of DMBT1 expression and localization, pointing to a chronological order of events. Here we report on the investigation of DMBT1 in breast cancer in order to test whether these principles might also be attributable to other tumor types. Comprehensive mutational analyses did not uncover unambiguous inactivating DMBT1 mutations in breast cancer. Expression analyses in the human and mouse mammary glands pointed to the necessity of DMBT1 induction. While age-dependent and hormonal effects could be ruled out, 9 of 10 mice showed induction of Dmbt1 expression after administration of the carcinogen 7,12-dimethybenz(alpha)anthracene prior to the onset of tumorigenesis or other histopathological changes. DMBT1 displayed significant up-regulation in human tumor-flanking tissues compared to in normal breast tissues (P < 0.05). However, the breast tumor cells displayed a switch from lumenal secretion to secretion to the extracellular matrix and a significant down-regulation compared to that in matched normal flanking tissues (P < 0.01). We concluded that loss of expression also is the predominant mode of DMBT1 inactivation in breast cancer. The dynamic behavior of DMBT1 in lung carcinoma is fully reflected in breast cancer, which suggests that this behavior might be common to tumor types arising from monolayered epithelia.


Subject(s)
Agglutinins , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carcinogens/metabolism , Down-Regulation/genetics , Receptors, Cell Surface/genetics , Adult , Aged , Aged, 80 and over , Animals , Breast Neoplasms/metabolism , Calcium-Binding Proteins , Cell Line, Tumor , DNA Mutational Analysis/methods , DNA-Binding Proteins , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Genes, Tumor Suppressor , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mammary Glands, Human/drug effects , Mammary Glands, Human/pathology , Mammary Glands, Human/physiology , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Middle Aged , Mutation/drug effects , Mutation/genetics , Receptors, Cell Surface/biosynthesis , Receptors, Cell Surface/metabolism , Tumor Suppressor Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...